Novel Classes of Antibiotics or More of The Same?: Review

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

British Journal of DOI:10.1111/j.1476-5381.2011.01250.

BJP Pharmacology
www.brjpharmacol.org

Themed Issue: Respiratory Pharmacology


Correspondence
Anthony RM Coates, Medical

REVIEW bph_1250 184..194


Microbiology, Centre for
Infection, Department of Clinical
Sciences, St George’s, University
of London, Cranmer Terrace,
Novel classes of antibiotics London SW17 ORE, UK. E-mail:
acoates@sgul.ac.uk
----------------------------------------------------------------

or more of the same? Keywords


novel antibiotics; bacterial
infections; antibiotic resistance;
Anthony RM Coates1, Gerry Halls2 and Yanmin Hu1 genomics; non-culturable
bacteria; bacteriophages;
1 non-multiplying bacteria;
Medical Microbiology, Centre for Infection, Department of Clinical Sciences, St George’s,
multiplying bacteria
University of London, Cranmer Terrace, London, UK and 2Medical Marketing Services, ----------------------------------------------------------------
Beaconsfield, Buckinghamshire, UK Received
29 December 2010
Revised
13 January 2011
Accepted
14 January 2011

The world is running out of antibiotics. Between 1940 and 1962, more than 20 new classes of antibiotics were marketed.
Since then, only two new classes have reached the market. Analogue development kept pace with the emergence of resistant
bacteria until 10–20 years ago. Now, not enough analogues are reaching the market to stem the tide of antibiotic resistance,
particularly among gram-negative bacteria. This review examines the existing systemic antibiotic pipeline in the public
domain, and reveals that 27 compounds are in clinical development, of which two are new classes, both of which are in
Phase I clinical trials. In view of the high attrition rate of drugs in early clinical development, particularly new classes and the
current regulatory hurdles, it does not seem likely that new classes will be marketed soon. This paper suggests that, if the
world is to return to a situation in which there are enough antibiotics to cope with the inevitable ongoing emergence of
bacterial resistance, we need to recreate the prolific antibiotic discovery period between 1940 and 1962, which produced 20
classes that served the world well for 60 years. If another 20 classes and their analogues, particularly targeting gram-negatives
could be produced soon, they might last us for the next 60 years. How can this be achieved? Only a huge effort by
governments in the form of finance, legislation and providing industry with real incentives will reverse this. Industry needs to
re-enter the market on a much larger scale, and academia should rebuild its antibiotic discovery infrastructure to support this
effort. The alternative is Medicine without effective antibiotics.

LINKED ARTICLES
This article is part of a themed issue on Respiratory Pharmacology. To view the other articles in this issue visit
http://dx.doi.org/10.1111/bph.2011.163.issue-1

Abbreviations
DNA, deoxyribonucleic acid; HAP, Hospital acquired pneumonia; HIV/AIDS, human immunodeficiency/ acquired
immunodeficiency syndrome; MRSA, methicillin resistant Staphylococcus aureus; RNA, ribonucleic acide; spp, species; TB,
tuberculosis; VAP Ventilator associated pneumonia

Introduction (Watson, 2008). In the short term, according to the Infectious


Diseases Society of America (IDSA, 2010) at least another 10
The future of modern medicine depends upon effective anti- antibiotics, which are active against superbugs, are required
biotics (So et al., 2010). The world produced more than 20 to reach the market within the next 10 years. In the longer
novel classes of antibiotics between 1930 and 1962 (Coates term, novel classes of antibiotics will be needed, but how
et al., 2002; Powers, 2004). Since then, only two new classes many? The answer to this question is unknown. However, on
of antibiotics have been marketed (Butler and Buss, 2006; the basis that most classes of antibiotics now have substantial
Hair and Kean, 2007; Zappia et al., 2007). Numerous ana- resistance problems, at least for some species of bacteria, a
logues of existing classes have reached the market in this time new class, together with its analogues, might remain useful
period. Meanwhile, multi-drug-resistant bacteria (superbugs) for 50 years. This would mean that the 20 classes which
have emerged throughout the world (Levy and Marshall, entered the market between 1930 and 1960 lasted for an
2004), and now half the deaths from clinical infection in average of 50 years. This assumption would lead to the con-
Europe are associated with multi-drug-resistant bacteria clusion that it is likely that the world will need a further 20

184 British Journal of Pharmacology (2011) 163 184–194 © 2011 The Authors
British Journal of Pharmacology © 2011 The British Pharmacological Society
New antibiotic classes are urgently needed
BJP
New Classes tially over the past two to three decades; now, only five
remain active in the field (Boucher et al., 2009). Governments
Marketed Needed
did not increase funding for antibiotic discovery, including
education in this field, fast enough to cope with the rise in
antibiotic resistance (House of Lords, 1998). This is a major
[-----20-----] [--2--] [-------------20?-----------][----------- 20?----------------] concern for many countries. Meanwhile, regulatory require-
ments increased, which has elevated the costs of developing
Year:
drugs, and the profits from competing products led pharma-
ceutical companies away from the antibiotic field.
1940—50—60—70—80—90—2000—10—20—30—40—50—60—70—80—90—2100—10

Why is the scope for analogue


Figure 1
development limited?
The number of new classes of antibiotics which have reached the
Table 1 shows the number of analogues which have been
market, and predictions of novel classes of antibiotics which are
developed in each class of antibiotics. Although there is more
needed during the next 100 years.
scope for further analogue development, it seems that such
an approach is more feasible for some classes than others,
novel classes of antibiotics to support modern medicine cephalosporins, penicillins and quinolones being the most
during the next 50 years (see Figure 1). Some bacterial infec- successful, as there are sites in the scaffolds for easy modifi-
tions, such as gram-negatives, are already very difficult to cation. However, new analogues based on one core eventually
treat, for example, metallo-b-lactamase producing pathogens become too difficult or too expensive to make, particularly
which neutralize carbapenems (Kumarasamy et al., 2010), when faced with new antibiotic resistance mechanisms.
while others, such as gram-positive Staphylococcal infections
like methicillin-resistant Staphylococcus aureus (MRSA) are still New classes
susceptible to a range of old and new antibiotics (Boucher Can we produce new classes of antibiotics many times faster
et al., 2009; Walkey et al., 2010). Thus, the need is critical for than we have achieved over the past 50 years? Although only
some gram-negatives, but less so for gram-positives. However, two new classes of antibiotics were marketed in the past 10
the world’s capacity for antibiotic discovery is already falling years, this shows it is still possible to discover and market new
behind the rate of emergence of bacterial resistance, and so, is classes. It also suggests that the reason for the lack of new
it likely that another 20 new classes of antibiotics will reach class development between 1962 and 2000 was that pharma-
the market within 50 years, and perhaps, a further 20 classes ceutical companies were concentrating on analogue develop-
between 50 and 100 years from now? Figure 1 depicts a line ment, perhaps because the toxicity risks associated with
drawing of the number of new classes that reached the analogues is lower than that for new classes. Whether it will
market between the 1940s and 1960s, the two new classes be possible to increase antibiotic class discovery to the level it
since 2000, and projections into the future of 20 new classes was between 1940 and 1960, is another matter. It may be
for the next 50 years, and 20 further classes required to difficult to do so, based upon the possibility that most of the
support medicine in 50–100 years from now. Although these easy-to-discover antibiotics have already been marketed.
are only projections, they are drawn upon past performance Also, the workforce in companies and in the university sector
of the pharmaceutical industry and upon the relentless emer- need to be re-skilled in microbiology and pharmacology, with
gence of antibiotic resistant bacteria. Whether or not the particular emphasis on antibiotic discovery, and this will take
actual numbers of new classes required proves to be accurate, decades to achieve and will be very expensive. Most govern-
and whether the goal of 20 more classes in the next 50 years ment funding concentrates on the pathogenesis of infectious
is achievable, there is clearly an urgent need for new classes of disease, and while this is important, if new antibiotics are to
antibiotics. However, the production of only two new classes emerge from the university sector, a higher proportion of the
of antibiotics in the last 50 years suggests that it may be very funding must be allocated to antibiotic discovery and devel-
difficult to produce enough new classes of compounds to opment. Thus, to generate the number of new antibiotics
support modern medicine during the next 50 years. which are needed over the next 100 years, substantially
increased funding from industry and from government will
The issues be required, and should be focused upon antibiotic discovery,
Antibiotic discovery is the key to the continuance of modern with less funding for microbial pathogenesis.
medicine (So et al., 2010). Why is the pharmaceutical indus- The emergence of antibiotic resistance is at the heart of
try producing fewer new antibiotic classes now, when the the gradual demise of the antibiotic era. Can we produce
need is increasing? The reason for the lack of new classes is antibiotics which induce resistance at a lower rate than exist-
not clear, but the following will be discussed later in this ing antibiotics? This is a key question and will be addressed
paper: there may be a shortage of new metabolic targets further in the section on antibiotic resistance below.
(Becker et al., 2006). Marketing of analogues is more finan- The aims of this review are to discuss the advantages and
cially feasible than developing new classes (Devasahayam disadvantages of continuing antibiotic discovery in the tra-
et al., 2010), and it used to be thought that current antibiotics ditional way, namely, more of the same, meaning analogue
would last almost indefinitely (reviewed in Coates et al., development using the Fleming method. The role of the
2002). The numbers of major pharmaceutical companies relentless emergence of antibiotic resistance to all antibiotics
which are engaged in antibiotic discovery decreased substan- will also be discussed, as well as possible ways to design

British Journal of Pharmacology (2011) 163 184–194 185


ARM Coates et al.
BJP
Table 1
Main classes of marketed and withdrawn antibiotics

Class examples

b-Lactams
Penicillins
Penicillin G, penicillin V, methicillin, oxacillin, cloxacillin, dicloxacillin, nafcillin, ampicillin, amoxicillin, carbenicillin, ticarcillin,
mezlocillin, piperacillin, azlocillin, temocillin
Cephalosporins
First generation Cephalothin, cephapirin, cephradine, cephaloridine, cefazolin
Second generation Cefamandole, cefuroxime, cephalexin, cefprozil, cefaclor, loracarbef, cefoxitin, cefmetazole
Third generation Cefotaxime, ceftizoxime, ceftriaxone, cefoperazone, ceftazidime, cefixime, cefpodoxime, ceftibuten, cefdinir
Fourth generation Cefpirome, cefepime
Fifth generation1 Ceftaroline2, ceftobiprole3
Carbapenems
Imipenem, meropenem, doripenem
Monobactams
Aztreonam
b-Lactamase inhibitors
Clavulanate, sulbactam, tazobactam
Aminoglycosides
Streptomycin, neomycin, kanamycin, paromomycin, gentamicin, tobramycin, amikacin, netilmicin, spectinomycin, sisomicin, dibekacin,
isepamicin
Tetracyclines
Tetracycline, chlortetracycline, demeclocycline, minocycline, oxytetracycline, methacycline, doxycycline, tigecycline
Rifamycins
Rifampicin (also called rifampin), rifapentine, rifabutin, bezoxazinorifamycin, rifaximin
Macrolides
Erythromycin, azithromycin, clarithromycin
Ketolides
Telithromycin
Lincosamides
Lincomycin, clindamycin
Glycopeptides
Vancomycin, teicoplanin, telavancin
Lipopeptides
Daptomycin
Streptogramins
Quinupristin, dalfopristin, pristinamycin
Sulphonamides
Sulphanilamide, para-aminobenzoic acid, sulfadiazine, sulfisoxazole, sulfamethoxazole, sulfathalidine
Oxazolidinones
Linezolid
Quinolones
Nalidixic acid, oxolinic acid, norfloxacin, pefloxacin, enoxacin, ofloxacin/levofloxacin, ciprofloxacin, temafloxacin, lomefloxacin,
fleroxacin, grepafloxacin, sparfloxacin, trovafloxacin, clinafloxacin, gatifloxacin, moxifloxacin, sitafloxacin
Others
Metronidazole, polymyxin B, colistin, trimethoprim

1
Spectrum combines third generation gram-negatives with methicillin resistant Staphylococcus aureus (MRSA) and multidrug-resistant
Streptococcus pneumoniae.
2
Pending Federal Drugs Administration review.
3
Marketed in Canada and Switzerland; now withdrawn.

186 British Journal of Pharmacology (2011) 163 184–194


New antibiotic classes are urgently needed
BJP
antibiotics which lead to a lower rate of emergence of resis- Another potential new class, PDF inhibitors, has been aban-
tance. The merits of trying to discover new classes of antibi- doned by a number of companies over the past decade
otics will be discussed, including ways in which this could be because of resistance emergence.
achieved. Finally, the future direction of antibiotic discovery For gram-negatives, the non-b-lactamase b-lactamase-
will be covered, with advantages and disadvantages of differ- inhibitor from Novexel has broad coverage against class A, C
ent routes. and some class D, such as OXA-48, but not those of Acineto-
bacter; it has no activity against metallo-b-lactamases. Merck
& Co. also have a similar analogue, but there is less informa-
More of the same tion available in public domain about this molecule.
The siderophore monobactams from Basilea and Pfizer
The traditional, and the most successful way of making anti- have similar structures but seem to have different activities,
biotics has been to find a natural product (Singh and Barrett, especially against Acinetobacter spp. The combination of
2006) or, in a few cases, a chemically synthesized one BAL30072 with meropenem seems to cope with many of the
(Fernandes, 2006) by screening for activity against a bacterial b-lactam-resistance mechanisms in gram-negatives: this is an
culture which is in log phase growth. For example, penicillin example of more than just inhibiting resistance mechanisms,
(Fleming, 1929) was discovered by Alexander Fleming by but of broadening the spectrum. However, none of the devel-
observing the antibacterial effect of the Penicillium fungus. opments is a panacea. Even the new class from Anacor/
This discovery led to the creation of numerous analogues, GlaxoSmithKline has gaps, especially Acinetobacter spp. The
many of which were marketed (see Table 1). The pharmaceu- neoglycoside from Achaogen is immune to nearly, but not all,
tical industry was able to build the analogues in such a way the aminoglycoside modifying enzymes found in Enterobac-
that they were active against penicillin-resistant bacteria. The teriaceae but is not active against Pseudomonas aeruginosa.
advantage of this route is that analogues tend to have a It should be noted that in Table 2, the indications for
similar solubility, protein binding and toxicity as the parent products at preclinical, Phase I and Phase II are largely
compound. A further advance that was made was to add an guesswork
anti-bacterial resistance factor to the antibiotic, which ren-
dered it active in the patient. An example of this approach is
the addition of clavulanic acid, a beta-lactamase inhibitor, to Resistance – the antibiotic slayer
amoxicillin, which is marketed as Augmentin. The disadvan-
tage of these routes of development is that there is a limit to The Achilles heel of antibiotics is resistance development of
the number of analogues which can be made from a single target pathogenic bacteria. Within 2 years of marketing, resis-
chemical core, or ones which can counteract bacterial resis- tance is usually observed, even to new classes of compounds
tance mechanisms, and eventually, bacteria can evolve resis- (Bax et al., 1998). When the proportion of bacteria, which
tance beyond the scope of even the most ingenious medicinal cause specific types of clinical infection, rises to 20% or more,
chemist. Other discoveries followed that of penicillin, the the antibiotic becomes poorly effective for that indication
most notable of which, in terms of analogue development, and may be withdrawn. This is a unique feature of anti-
was cephalosporins (see Table 1). Even today, cephalosporin microbial drugs, namely that they become ineffective over
analogues are being developed with anti-superbug activity, time, something which does not affect other drug groups
one of which, ceftaroline, is in pre-registration (see Table 2). such as cardiovascular, central nervous system and anti-
Clearly, there is some scope for more analogue development, inflammatories. However, some bacteria mutate slowly, while
and a new trimethoprim derivative, iclaprim, was recently others do so faster or acquire resistance elements from other
rejected by the Federal Drugs Administration (FDA) for failure species. For example, some strains of Neisseria meningitidis (du
to demonstrate non-inferiority. However, it is also clear that Plessis et al., 2008) still show only a reduced susceptibility to
some antibiotic families are more amenable to analogue penicillin, while for S. aureus (Chambers and Deleo, 2009)
development than others. For instance (Table 1), the most resistance to penicillin appeared in the early 1940s, shortly
prolific number of analogues has been made using the after its introduction into the market, and resistance to
cephalosporin and penicillin cores, but the quinolone and methicillin was recorded just 1 year after its introduction. In
aminoglycoside cores have also been very productive. Unfor- 2005 in the USA, it has been estimated that S. aureus caused
tunately, resistance arises to all these compounds, and thus 10 800 deaths, of which 5500 were due to MRSA (Klein et al.,
analogue development merely ‘buys time’ until the discovery 2007). The majority of hospital infections in USA are caused
of the next novel class. However, in our view, more analogues by the so-called ESKAPE pathogens (Rice, 2010), for which
are likely to be marketed in the next 20 years. Table 2 shows new antibiotics are urgently needed. These are Enterococcus
the antibiotics which are in development and which are faecium, S. aureus, Acinetobacter baumanii, Klebsiella pneumo-
accompanied by information in the public domain. It can be niae, P. aeruginosa and Enterobacter species. Gram-negative bac-
seen that although few new classes are represented, there are teria are particularly troublesome in this regard, and have
some significant improvements. Concern has to be noted become progressively resistant to each antibiotic class. Now
that pleuromutilins, which are new to human therapy but Enterobacteriaceae (Kumarasamy et al., 2010), which are
have been used in veterinary medicine for about 30 years, are resistant to carbapenem conferred by New Delhi metallo-b-
susceptible to the cfr plasmid-mediated resistance which lactamase 1, are being isolated from patients in several coun-
affects virtually all antibiotics that target the 50S ribosomal tries. Major efforts are being adopted by many countries to
subunit. In this review, we do not count pleuromutilins as a prevent such infections (Coates and Hu, 2008) by improve-
new class because they have been widely used for decades. ments in infection control in hospitals, withdrawal of

British Journal of Pharmacology (2011) 163 184–194 187


Table 2
Antibacterial compounds in development BJP
Class Product Spectrum Iv/oral Indications Phase Company (Licensor)

Glycopeptide Dalbavancin Gram-positive (excluding VRE) IV; once-weekly cSSTI Phase III Durata Therapeutics
(Pfizer)
Glycopeptide Telavancin Gram-positive (excluding VRE) IV cSSTI and HAP Marketed for SSTI in Astellas (Theravance)
USA; HAP approval
ARM Coates et al.

stalled at FDA
Glycopeptide Oritavancin Gram-positive (including VRE) IV; single dose cSSTI Phase III The Medicines
treatment Company (Lilly)
Cephalosporin Ceftaroline Gram-positive and gram-negative IV SSTI, CAP Pre-registration Forest (Cerexa)
excluding ESBLs etc. and
non-fermenters
Glycopeptide- TD-1792 Gram-positive IV SSTI, HAP Phase IIa Theravance
cephalosporin hybrid
Ketolide Cethromycin Gram-positive and respiratory tract Oral and IV Community-acquired RTIs Phase III Advanced Life Sciences

188 British Journal of Pharmacology (2011) 163 184–194


infection pathogens biothreat pathogens (Abbott)
Ketolide EDP-420 Gram-positive and respiratory tract Oral Community-acquired RTIs Phase II/III Enanta (Shionogi)
infection pathogens
Fluoroketolide Solithromycin Gram-positive and respiratory tract IV/oral Community-acquired RTIs Phase I/II Cempra (Optimer)
infection pathogens biothreat pathogens
Pleuromutilin BC-3781 Gram-positive and respiratory tract Oral IV Community-acquired RTIs Phase II Nabriva
infection pathogens and SSTIs Preclinical
Peptide deformylase GSK1322322 Gram-positive and Respiratory tract Oral Community-acquired RTIs Phase I GlaxoSmithKline
inhibitor (new class) pathogens and SSTIs
Oxazolidinone Torezolid Gram-positive (including linezolid- and IV/oral cSSTI Phase IIa Trius (DongA)
daptomycin resistant strains)
Oxazolidinone Radezolid Gram-positive (including linezolid- and IV/oral SSTI Phase IIa Rib-X
daptomycin resistant strains)
Oxazolidinone PNU-100480 TB Oral TB Phase I Pfizer
FabI Inhibitor AFN-1252 Staphylococci Oral Staph infections Phase I Affinium
(new class)
FabI Inhibitor MUT056399 Staphylococci IV Staph infections Phase I FAB Pharma
(new class)
Aminomethylcycline PTK0796 Gram-positive, RTI and SSTI pathogens IV/oral cSSTI, CAP Phase III Novartis (Paratek)
(tetracycline)
Fluoroquinolone Delafloxacin Broad-spectrum including IV/oral cSSTI, Phase III Rib-X
fluoroquinolone-resistant MRSA
Fluoroquinolone Finafloxacin Broad-spectrum; enhanced activity at Merlion
acid pH
Table 2
Continued.

Class Product Spectrum Iv/oral Indications Phase Company (Licensor)

Fluoroquinolone JNJ-Q2 Enhanced gram-positive activity including IV/oral Phase I J&J


fluoroquinolone-resistance-resistant MRSA
Fluorocycline (tetracycline) TP-434 Broad gram-positive, -negative and anaerobic IV/oral cSSTI, cIAI Phase I Tetraphase
activity including Acinetobacter but not P.
aeruginosa, less active versus Proteae and
some K. pneumoniae
Aminoglycoside ACHN-490 MDR enterobacteriaceae and S. aureus, IV cUTI, cIAI Phase I Achaogen
including aminoglycoside-resistant and
metallo-ß-lactamase producers
leucyl-tRNA synthase GSK2251052 MDR enterobacteriaceae and P. aeruginosa IV Anacor/GSK
inhibitor (new class)
Penicillin CXA-101 MDR P. aeruginosa and susceptible IV cUTI Phase II Cubist
enterobacteriaceae
Penicillin/b-lactamase- CXA-101 /tazobactam MDR P. aeruginosa and enterobacteriaceae, IV cUTI, cIAI Phase I Cubist
inhibitor (CXA-201) excluding metallo-ß-lactamases VAP
Non-b-lactam b-lactamase- NXL104 Class A, C and some class D b-lactamases, AstraZeneca (Novexel)
inhibitor (new class) including OXA-48
Cephalosporin/b- Ceftaroline/ NXL104 MRSA and MDR enterobacteriaceae, excluding IV cUTI, SSTI, CAP Phase II ready AstraZeneca/Forest
lactamase-inhibitor metallo-ß-lactamases
Cephalosporin/b- Ceftazidime/ NXL104 MDR P. aeruginosa and enterobacteriaceae, IV cUTI, SSTI, VAP Phase III ready AstraZeneca/Forest
lactamase-inhibitor excluding metallo-ß-lactamases
Non-b-lactam MK-7655 Class A and C b-lactamases Merck & Co
b-lactamase-inhibitor
(new class)
Carbapenem/b- Imipenem/ MK-7655 MDR P. aeruginosa and enterobacteriaceae, IV cUTI, cIAI, HAP/VAP Phase II ready Merck & Co
lactamase-inhibitor excluding metallo-ß-lactamases;
Acinetobacter
Sulfactam (siderophore BAL30072 MDR P. aeruginosa Acinetobacter including IV Preclinical Basilea
monobactam) metallo-ß-lactamases and
enterobacteriaceae
Monobactam/ carbapenem BAL30072/ meropenem Most MDR gram-negatives including resistant IV cUTI, Hospital-acquired Preclinical Basilea
Enterobacteriaceae and anaerobes cIAI, VAP
Monocarbam (siderophore MC-1 MDR P. aeruginosa including IV cUTI, Hospital-acquired Preclinical Pfizer
monobactam) metallo-ß-lactamases S. maltophilia and cIAI, VAP
enterobacteriaceae
New antibiotic classes are urgently needed

These data are based upon information in the public domain.


Products shaded blue = primarily gram-positive; unshaded = broad(ish)-spectrum; shaded red = MDR gram-negatives.
MDR, multidrug resistant; MRSA, methicillin-resistant Staphylococcus aureus.
BJP

British Journal of Pharmacology (2011) 163 184–194 189


ARM Coates et al.
BJP
vulnerable classes of antibiotics, restrictions in the use of Stage Compounds Marketed (prediction)*
antibiotics and the introduction of new vaccines.
No (new classes)
However, superbugs which are resistant to key classes of
antibiotics continue to emerge. Some antibiotics induce resis-
tance readily, for example, rifampicin (Lambert, 2005), while Phase I 11(2) 1(0)---------------------]
others, such as those which target the cell membrane, may do
Phase II 8(0) 2(0)---------------------]
so more slowly (Zhanel et al., 2008). The key mechanisms
of genetic resistance are (Coates et al., 2002): (i) bacteria Phase III 6(0) 3(0)---------------------]
can inactivate the antibiotic by producing, for example,
Preregistration 2(0) 2(0)
b-lactamase which degrades the b-lactam ring which is a key
part of penicillins and cephalosporins; (ii) reduce membrane
permeability to the antibiotic; (iii) increase the efflux of anti- Year 2010--------------------2015--------------------2020--------
biotic from the cell; (iv) overproduce the target enzyme; (v)
bypass the inhibited step; and (vi) alter the site of action of
the antibiotic. Some antibiotics, notably fluoroquinolones, Figure 2
induce the SOS response, which increases the error rate of The number of systemic compounds and classes in development and
DNA replication and speeds the development of resistance predictions of the number which will reach the market. Based on
(Da Re et al., 2009) estimates of drugs in development which are published in the public
In addition, before the onset of genetic resistance, bacte- domain. New classes are in brackets. *Assumptions: percent of com-
pounds which reach the market (CMR International 2009) – 6.25%
ria can survive antibiotic treatment by entering into a slow or
in Phase I, 25% in Phase II, 50% in Phase III and 75% in Pre-
non-multiplying state (Coates et al., 2002). It is thought that
registration. Phase II includes drugs which are in Phase I/II, II ready,
about 60% of all clinical infections contain bacteria in this II and IIa. Phase III includes drugs in II/III, III ready and III. Market
state (Coates and Hu, 2008). Commensal bacteria, for predictions are shown with – which indicates a range of years due to
example, those which naturally live on the skin, in the current uncertainty of the length of time – which is required to
mouth, nose and intestines contain large numbers of complete Phase III trials, for example for hospital-acquired
antibiotic-resistant organisms, and these may be a source of pneumonia/ventilator-associated pneumonia.
antibiotic-resistance markers for pathogenic bacteria (Gillings
et al., 2008). Also, about half of all antibiotics which are used
the antibiotics in clinical development, which appear in
each year in the world are consumed by animals. This is also
Table 2 and Figure 2, are based upon information which is in
thought to be a source of antibiotic resistance in humans,
the public domain. However, this may be an underestimate,
although this is disputed by some (Soulsby, 2008). Recently
because many companies do not publish data in this area.
(van Cleef et al., 2010), MRSA has been found in half of
Figure 2 shows that there are at least 27 anti-bacterial com-
the pig farms in Europe and has spread to pig farmers and
pounds which are in clinical development, of which 11 are in
their families, but has not, as of yet, spread into the general
Phase I clinical trials, 8 in Phase II, but only 6 in Phase III and
population.
2 at the pre-registration stage. Of the antibiotics in clinical
These data suggest that antibiotic resistance should not be
development, two belong to new classes. There are no antibi-
considered, particularly in gram-negatives, to be isolated to a
otics against the major gram-negative pathogens K. pneumo-
small number of superbugs. Rather, it is part of a much larger
niae, P. aeruginosa and A. baumanii in Phase IIb and III.
picture, namely the whole of the bacterial kingdom which
Furthermore, there are only a few compounds against these
seems to operate cooperatively, horizontally transferring anti-
pathogens in earlier stages of development. Most of these
biotic resistance containing DNA between different species.
compounds are analogues of existing marketed antibiotics.
Also, the resistant bacteria are fit to be able to survive and
Because there are no new class in the later stages of develop-
persist for a long period of time, even though no antibiotic
ment, Phase II, III and pre-registration (Figure 2), there may be
selective pressure is present (Andersson, 2006)
no new classes in the market in the short term. Although there
are two potential new class compounds in early clinical Phase
I development, the high attrition rate, particularly for new
Novel classes of antibiotics
classes, means that the odds are against these compounds
reaching the market, and it is possible that no new classes will
Novel classes of antibiotics are urgently needed for the future.
reach the market within 10 years. In the longer term, during
The two new classes of antibiotics which have been intro-
the next 20 years, the likelihood of discovering 20 novel
duced into the market, oxazolidinone (linezolid by Pfizer) and
classes including many broad spectrum antibiotics, similar to
cyclic lipopeptide (daptomycin by Cubist) are active against
the achievements in the 1940–1960s, seems to be remote,
gram-positive bacteria, such as MRSA, but there are no new
particularly for multi-drug-resistant gram-negatives.
classes in Phase II or III clinical trials, and none in the pre-
registration stage (see Figure 2). There are two new classes in
Phase I and a small number in the pre-clinical and discovery
phases, for example, a new class which targets type IIA topoi- Why is there a shortage of new classes
somerases with a new mechanism of action (Bax et al., 2010). of antibiotics in development?
It is difficult to estimate the number of drugs in preclinical
development, because most are not published, so no attempt The risk and cost of developing a new class is considerably
has been made to do so in this review. It should be noted that greater than that of an analogue. For example, the starting

190 British Journal of Pharmacology (2011) 163 184–194


New antibiotic classes are urgently needed
BJP
point for an analogue is likely to give rise to compounds Future direction
which are soluble and are not toxic, on the grounds that the
parent molecule has these characteristics. Toxic side effects If bacteria continue to develop new resistance mechanisms,
are a common cause of failure of a compound to proceed past and if the pharmaceutical industry produces novel classes of
Phase I clinical trials. In contrast, a new class of compound antibiotics at the existing rate, the future for medicine is
has unknown toxic potential, and may have chemico- bleak. In our view, it is unlikely that the industry will dra-
physical features which make them unsuitable for drug matically increase the rate of production of new classes of
development. antibiotics. This means that the present global antibiotic
However, there may be a more fundamental reason why discovery process is probably unsustainable in the long
so few new classes have reached the market during the past term.
50 years. Simply, it may be more difficult to find new classes
which are broad spectrum, than it was in the golden age of
antibiotic discovery. It has been suggested (Becker et al.,
2006) that anti-bacterial classes have been developed and How can we improve this situation?
marketed already against all the main metabolic pathways,
and that there are no more available. In other words, it is Reduce the rate of emergence of resistance
possible that all the classes of broad-spectrum antibacterials The most important step would be to break the cycle of
have been discovered. If this were to be the case, the antibi- marketing a new antibiotic, followed by resistance arising
otic era is likely to fade away, sooner rather than later. within a few years. Tuberculosis (TB) chemotherapy (Mitchi-
Another blow to the antibiotic discovery field is the con- son, 2005) has addressed this issue by using combinations of
siderable investment which the pharmaceutical industry antibiotics which reduce the rate of emergence of resistance
invested in the genomics approach (Brotz-Oesterhelt and in the target pathogen. However, multi-drug-resistant TB is
Sass, 2010) that failed to deliver a new class into the market. on the increase, and so, while this approach can slow the
This approach uses bacterial DNA sequence data to predict emergence of resistance, it cannot stop it. However, combi-
enzymatic pathways which can be inhibited by novel classes nation therapy is an approach which could be applied more
of compounds. widely for the treatment of pyogenic bacteria. A potential
The recent difficulties with registration of antibiotics are disadvantage of this approach is that it could increase the rate
also likely to suppress the enthusiasm of large pharmaceutical of resistance emergence in normal bacterial flora, and these
companies to enter the field. For example, the FDA in the bacteria might then transfer resistance to pathogens.
USA, having agreed to the protocol for telavancin hospital- Included in this combination approach is the use of inhibi-
acquired pneumonia (HAP)/ventilator-associated pneumonia tors such as clavulanic acid which block the action of bacte-
(VAP) studies with clinical response as the primary endpoint, rial b-lactamase (Brogden et al., 1981). Another potential way
the agency requested additional data and analyses to support to deal with this issue is to develop antibiotics which induce
an evaluation of all-cause mortality as the primary efficacy resistance at a lower rate than existing antibiotics. For
endpoint. As a result, the company may have to conduct new example, some compounds which target complex bacterial
studies. Initial applications for dalbavancin and iclaprim systems such as the membrane (Tenover, 2006; Hurdle et al.,
have also been rejected by the FDA. 2011), may induce resistance less readily than those such as
The requirement of the FDA for a smaller non-inferiority rifampicin (Falagas et al., 2007), which inhibit single enzymes
margin and to restrict nosocomial pneumonia trials to VAP is like RNA polymerase. Presumably, because synthesis of the
unlikely to result in new agents for multidrug resistant (MDR) membrane requires so many enzymes, bacteria cannot easily
gram-negative pneumonia. For a non-inferiority margin of produce mutants with altered membranes. While this is a
7%, the cost of a HAP/VAP study would total $600 million plausible hypothesis, resistance to daptomycin, a drug which
and take 8–10 years for enrolment, even using hundreds of targets bacterial membranes, is already occurring (Sakoulas
centres. This would double if the trial were to be VAP only. et al., 2006).
Further regulatory difficulties are envisaged that will discour-
age the development of MDR-gram-negative antibiotics.
With no standard therapy, non-inferiority studies are not Increase the rate of production of new
possible; historical controls are considered irrelevant and classes of antibiotics
superiority studies are not ethical as patients cannot be Historically, natural compounds have been the source of
randomized to ineffective therapy for infections with high most new classes of antibiotics. The search for new classes
mortality. needs to be intensified in both natural and chemical potential
Most large pharmaceutical companies have now left the sources and broadened to include novel approaches. For
antibiotic discovery field. This has led to de-skilling of the example, an actinomyte from the deep ocean has recently
workforce in the industry over the past 30 years. In parallel, been found which produces a novel antibiotic called abysso-
many universities have closed academic departments mycin (Riedlinger et al., 2004). Other sources such as plants
which underpin antibiotic discovery and development. The (Mitscher et al., 1987), reptiles (Wang et al., 2008) and
net effect of the de-skilling in antibiotic discovery world- mammals (Flores-Villasenor et al., 2010) are also being
wide, compared with the 1940–1960s, is that it will take searched. Another possible source of new classes is non-
decades to rebuild the teaching and training which is cultivable bacteria (Daniel, 2004), which constitute the
needed to support the discovery of numerous new classes of majority of bacterial species. It has been suggested that the
antibiotics. antibiotic-producing genes of these bacteria could be

British Journal of Pharmacology (2011) 163 184–194 191


ARM Coates et al.
BJP
expressed in cultivable bacteria, and so new antibiotic classes Conclusion
might be produced. Chemical libraries are also being screened
with the hope of finding new classes of anti-bacterials. In The existing antibiotic pipeline and the infrastructure which
addition, the genomics approach (Payne et al., 2007) in underpins it, is not sufficient to cope with the emergence of
which essential enzyme pathways in bacteria are targeted by resistance worldwide, particularly among gram-negative bac-
inhibitors, may provide new classes, although none have teria. If the antibiotic discovery process continues at the same
been marketed so far. A further new method which targets rate as at present ‘More of the same’, the antibiotic era will
non-multiplying bacteria (Hu et al., 2010) may also have end within a few decades, at least for many gram-negative
potential, and a new class topical compound derived in this infections.
way is now in Phase III clinical trials, but no new compounds Potential ways forward include the use of combinations of
have been marketed using this approach thus far. antibiotics to reduce the rate of emergence of resistance.
A crisis of this magnitude clearly needs government Concentrating on the discovery of compounds that target
incentives, although money alone will be insufficient. There complex bacterial systems, such as membranes, could also
needs to be a political appreciation globally that infections reduce resistance emergence.
(including those of the lower respiratory tract, human immu- In the long term, many more new classes of antibiotics are
nodeficiency virus/acquired immunodeficiency syndrome, needed. This will require the intervention of governments
diarrhoea, TB, malaria), of which antibiotic-resistant worldwide, with increased grants, subsidies, tax incentives
microbes are an important part, kill more people than heart and an increase in the unit price of new classes of antibiotics.
attacks or strokes (Lopez and Mathers, 2006). The current Universities, charities and regulatory bodies will also need to
effects of climate change on health are more difficult to judge, change in order to encourage antibiotic discovery.
but if predictions are accurate, deaths due to infections such
as diarrhoeal disease may increase as a result of
climate*induced changes (IPCC, 2007). In addition, it needs
to be understood that antibiotics are not like other groups of Acknowledgements
drugs such as anti-inflammatory agents as they become
redundant in a few decades and so have to be continually AC and YH would like to acknowledge financial support from
replaced. Many governments already provide financial incen- the Medical Research Council, European Commission and
tives, such as grants to academia and industry in the field of the Burton Trust.
infectious disease. However, thus far, an insufficient propor-
tion of these grants have been allocated to academia and
industry to stimulate the generation of enough new classes of
antibiotics with which to counteract the tide of antibiotic Conflicts of interests
resistance.
The pharmaceutical industry needs to be encouraged to AC is founder, director and shareholder of Helperby Thera-
return to the antibiotic discovery arena. Only the prospect peutics Group Ltd which makes new antibiotics targeted at
of profit from a marketed antibiotic product will persuade non-multiplying bacteria. AC and YH receive grants from
them to do this. The simplest way forward would be for Helperby Therapeutics.
governments to agree to a cost per unit for novel antibiotics
which is comparable to anti-cancer drugs. Patent protection
should be specifically extended for antibiotics on the
grounds so that this will encourage companies to discover
novel antibiotics, and the enhanced price should reduce References
the usage of the product and should extend the life of the
antibiotic. Andersson DI (2006). The biological cost of mutational antibiotic
resistance: any practical conclusions? Curr Opin Microbiol 9:
Universities should be encouraged to rebuild their antibi-
461–465.
otic discovery sectors and to replace lost skills in this field.
Clearly this will take decades, but antibiotic discovery is Bax RP, Anderson R, Crew J, Fletcher P, Johnson T, Kaplan E et al.
something that will need to be continued into the foreseeable (1998). Antibiotic resistance – what can we do? Nat Med 4:
future. 545–546.
The considerable resources of the charitable sector should Bax BD, Chan PF, Eggleston DS, Fosberry A, Gentry DR, Gorrec F
be channelled in part into antibiotic discovery for new classes et al. (2010). Type 11A topoisomerase inhibition by a new class of
of drugs against gram-negative bacteria, in a similar way to antibacterial agents. Nature 19: 935–940.
that for the TB and malaria programmes.
Becker D, Selbach M, Rollenhagen C, Ballmaier M, Meyer TF,
Infection control also has a role, as have better diagnostics
Mann M et al. (2006). Robust salmonella metabolism limits
and new vaccines. possibilities for new antimicrobials. Nature 16: 303–307.
A major change is now needed in the regulation of anti-
biotics. The rules are too restrictive and need to be relaxed for Boucher HW, Talbot GH, Bradley JS, Edwards JE, Gilbert D, Rice LB
this group of drugs because the world is running out of et al. (2009). Bad bugs, no drugs: no ESKAPE! An update from the
Infectious Diseases Society of America. Clin Infect Dis 48: 1–12.
antibiotics. For example, in pivotal clinical trials, specific
surrogate endpoints could be allowed, such as a microbiologi- Brogden RN, Carmine A, Heel RC, Morley PA, Speight TM,
cal endpoint, rather than a clinical endpoint. Avery GS (1981). Combination clavulanic acid. Drugs 22: 337–362.

192 British Journal of Pharmacology (2011) 163 184–194


New antibiotic classes are urgently needed
BJP
Brotz-Oesterhelt H, Sass P (2010). Postgenomic stategies in IDSA (2010). ID Physicians call for 10 new antibiotics by 2020.
antibacterial drug discovery. Future Microbiol 5: 1553–1579. Available at http://www.journals.uchicago.edu/doi/full/10.1086/
652237 (accessed 19 February 2010).
Butler MS, Buss AD (2006). Natural products – the future scaffolds
for novel antibiotics? Biochem Pharmacol 71: 919–929. IPCC (2007). Climate change 2007: synthesis report. Contribution
of working groups I, II and III to the fourth assessment report of
Chambers HF, Deleo FR (2009). Resistance Staph aureus to the intergovernmental panel on clinate change. In: Pachauri RK,
penicillin and methicillin. Nat Rev Microbiol 7: 629–641. Reisinger A (eds). Core Writing Team. IPCC: Geneva, p. 104.
van Cleef BA, Verkade EJ, Wulf MW, Buiting AG, Voss A, http://www.ipcc.ch/publicationsanddata/publicationsipccfourth
Huijsdens XW et al. (2010). Prevalence of livestock-associated MRSA assessmentreportsynthesisreport.htm.
in communities with high pig-densities in the Netherlands. PLoS Klein E, Smith DL, Laxminarayan R (2007). Hospitalizations and
ONE 5: e9385. deaths caused by methicillin-resistant Staphylococcus aureus,
Coates AR, Hu Y (2008). Targeting non-multiplying organisms as a United States, 1999–2005. Emerg Infect Dis 13: 1840–1846.
way to develop novel antimicrobials. Trends Pharmacol Sci 29: Kumarasamy KK, Toleman MA, Walsh TR, Bagaria J, Butt F,
143–150. Balakrishnan R et al. (2010). Emergence of a new antibiotic
Coates A, Hu Y, Bax R, Page C (2002). The future challenges facing resistance mechanism in India, Pakistan, and the UK: a molecular,
the development of new antimicrobial drugs. Nature Rev Drug biological and epidemiological study. Lancet Infect Dis 10:
Discovery 1: 895–910. 597–602.

CMR International (2009). Available at http://cmr.thomsonreuters. Lambert PA (2005). Bacterial resistance to antibiotics: modified
com/pdf/cmrfb-brochure.pdf (accessed February 2011). target sites. Adv Drug Deliv Rev 57: 1471–1485.

Da Re S, Garnier F, Guerin E, Campoy S, Denis F, Ploy MC (2009). Levy SB, Marshall B (2004). Antibacterial resistance worldwide:
The SOS response promotes qnrB quinolone-resistance determinant causes, challenges and responses. Nat Med 10: S122–S129.
expression. EMBO Rep 10: 929–933. Lopez AD, Mathers CD (2006). Measuring the global burden of
Daniel R (2004). The soil metagenome – a rich resource for the disease and epidemiological transitions: 2002–2030. Ann Trop Med
discovery of novel natural products. Curr Opin Biotechnol 15: Parasitol 100: 481–499.
199–204. Mitchison DA (2005). The diagnosis and therapy of tuberculosis
during the past 100 years. Am J Respir Crit Care Med 171: 699–706.
Devasahayam G, Scheld WM, Hoffman PS (2010). Newer
antibacterial drugs for a new century. Expert Opin Investig Drugs Mitscher LA, Drake S, Gollapudi SR, Okwute SK (1987). A modern
19: 215–234. look at folkloric use of anti-infective agents. J Nat Prod 50:
1025–1040.
Falagas ME, Bliziotis IA, Fragoulis KN (2007). Oral rifampin for
eradication of Staphylococcus aureus carriage from healthy and sick Payne DJ, Gwynn MN, Holmes DJ, Pompliano DL (2007). Drugs for
populations: a systematic review of the evidence from comparative bad bugs: confronting the challenges of antibacterial discovery. Nat
trials. Am J Infect Control 35: 106–114. Rev Drug Discov 6: 29–40.
Fernandes P (2006). Antibacterial discovery and development – the du Plessis M, von Gottberg A, Cohen C, de Gouveia L, Klugman KP
failure of success? Nat Biotechnol 24: 1497–1503. (2008). Neisseria meningitidis intermediately resistant to penicillin
and causing invasive disease in South Africa in 2001 to 2005. J Clin
Fleming A (1929). On the antibacterial action of cultures of a
Microbiol 46: 3208–3214.
penicillium, with special reference to their use in the isolation of
B.influenza. Br J Exp Path 10: 226–236. Powers JH (2004). Antimicrobial drug development – the past, the
present, and the future. Clin Microbiol Infect 4: 23–31.
Flores-Villasenor H, Canizalez-Roman A, Reyes-Lopez M, Nazmi K,
de la Garza M, Zazueta-Beltran J et al. (2010). Bactericidal effect of Rice LB (2010). Progress and challenges in implementing the
bovine lactoferrin, Lfcin, Lfampin and Lfchimera on antibiotic- research on ESKAPE pathogens. Infect Control Hosp Epidemiol 1:
resistant Staphylococcus aureus and Escherichia coli. Biometals 23: S7–S10.
569–578.
Riedlinger J, Reicke A, Zahner H, Krismer B, Bull AT, Maldonado LA
Gillings M, Boucher Y, Labbate M, Holmes A, Krishnan S, Holley M et al. (2004). Abyssomicins, inhibitors of the para-aminobenzoic
et al. (2008). The evolution of class 1 integrons and the rise of acid pathway produced by the marin Verrucosispora strain AB-
antibiotic resistance. J Bacteriol 190: 5095–5100. 18-032. J Antibiot (Tokyo) 57: 271–279.

Hair PI, Kean SJ (2007). Daptomycin: a review of its use in the Sakoulas G, Alder J, Thauvin-Eliopoulos C, Moellering RC,
management of complicated skin and soft-tissue infections and Eliopoulos GM (2006). Induction to daptomycin heterogeneous
Staphylococcus aureus bacteraemia. Drugs 67: 1483–1512. susceptibility in Staphylococcus aureus by exposure to vancomycin.
Antimicrob Agents Chemother 50: 1581–1585.
House of Lords (1998). Inquiry into antimicrobial resistance.
Commun Dis Rep CRD Wkly 24: 8. Singh SB, Barrett JF (2006). Empirical antibacterial drug discovery –
foundation in natural products. Biochem Pharmacol 30:
Hu Y, Shamaei-Tousi A, Liu Y, Coates A (2010). A new approach for
1996–1015.
the discovery of antibiotics by targeting non-multiplying bacteria: a
novel topical antibiotic for staphylococcal infections. PLoS ONE 5: So AD, Gupta N, Cars O (2010). Tackling antibiotic resistance. BMJ
e11818. Editorial 340: c2071.

Hurdle JG, O’Neill AJ, Chopra I, Lee RE (2011). Targeting bacterial Soulsby L (2008). The 2008 Garrod Lecture: antimicrobial
membrane function: an underexploited mechanism for treating resistance-animals and the environment. J Antimicrob Chemother
persistent infections. Nat Rev Microbiol 9: 62–75. 62: 229–233.

British Journal of Pharmacology (2011) 163 184–194 193


ARM Coates et al.
BJP
Tenover FC (2006). Mechanisms of antimicrobial resistance in Watson R (2008). Multidrug resistance responsible for half of deaths
bacteria. Am J Infect Control 34: S3–10, S64–73. from healthcare associated infections in Europe. BMJ 336: 1266.

Walkey AJ, O’Donnell MR, Wiener RS (2010). Linezolid versus Zappia G, Menendez P, Monache GD, Misiti D, Nevola L, Botta B
glycopeptide antibiotics for the treatment of suspected methicillin- (2007). The contribution of oxazolidinone frame to the biological
resistant Staphylococcus aureus nosocomial pneumonia: activity of pharmaceutical drugs and nature products. Mini Rev
a meta-analysis of randomized controlled trials. Chest 10: Med Chem 7: 389–409.
1556.
Zhanel GG, Trapp S, Gin AS, DeCorby M, Lagace-Wiens PR,
Wang Y, Hong J, Liu X, Yang H, Liu R, Wu J et al. (2008). Snake Rubinstein E et al. (2008). Dalbavancin and telavancin: novel
cathelicidin from Bungarus fasciatus is a potent peptide antibiotics. lipoglycopeptides for the treatment of Gram-positive infections.
PLoS ONE B3: e3217. Expert Rev Anti Infect Ther 6: 67–81.

194 British Journal of Pharmacology (2011) 163 184–194

You might also like